poxvirus-based active immunotherapy synergizes with ctla-4 … · cancer immunol immunother (2016)...

13
1 3 Cancer Immunol Immunother (2016) 65:537–549 DOI 10.1007/s00262-016-1816-7 ORIGINAL ARTICLE Poxvirus‑based active immunotherapy synergizes with CTLA‑4 blockade to increase survival in a murine tumor model by improving the magnitude and quality of cytotoxic T cells Susan P. Foy 1 · Stefanie J. Mandl 1 · Tracy dela Cruz 1 · Joseph J. Cote 1 · Evan J. Gordon 1 · Erica Trent 1 · Alain Delcayre 1,2 · James Breitmeyer 1 · Alex Franzusoff 1 · Ryan B. Rountree 1 Received: 5 November 2014 / Accepted: 23 February 2016 / Published online: 10 March 2016 © The Author(s) 2016. This article is published with open access at Springerlink.com co-stimulator (ICOS) protein was expressed predominantly on CD4 and CD8 effector T cells but not on regulatory T cells (T reg ). In contrast, mice left untreated or treated solely with CTLA-4 blockade harbored elevated ICOS + T reg , a phenotype associated with highly suppressive activity. In conclusion, poxvirus-based active immunotherapy induced robust tumor infiltration by highly efficient effector T cells. Combination with CTLA-4 immune checkpoint block- ade amplified this response resulting in synergistically improved efficacy. These hypothesis-generating data may help elucidate evidence of enhanced clinical benefit from combining CTLA-4 blockade with poxvirus-based active immunotherapy. Keywords Active immunotherapy · Poxvirus · Modified Vaccinia Ankara (MVA) · Cancer · Anti-CTLA-4 · Immune checkpoint blockade Abbreviations CEA Carcinoembryonic antigen CI Combination index CTL Cytotoxic T lymphocytes CTLA-4 Cytotoxic T lymphocyte-associated antigen-4 CV-301 Poxvirus-based active immunotherapy targeting CEA and MUC-1 DPECs Double-positive effector cells EECs Early effector cells HER-2 Human epidermal growth factor receptor 2 HER2 Modified extracellular domain of HER-2 and two additional T helper epitopes ICOS Inducible T cell co-stimulator IFN Interferon Abstract The dramatic clinical benefit of immune check- point blockade for a fraction of cancer patients suggests the potential for further clinical benefit in a broader can- cer patient population by combining immune checkpoint inhibitors with active immunotherapies. The anti-tumor efficacy of MVA-BN-HER2 poxvirus-based active immu- notherapy alone or in combination with CTLA-4 check- point blockade was investigated in a therapeutic CT26- HER-2 lung metastasis mouse model. MVA-BN-HER2 immunotherapy significantly improved the median over- all survival compared to untreated controls or CTLA-4 blockade alone (p < 0.001). Robust synergistic efficacy was achieved with the combination therapy (p < 0.01). Improved survival following MVA-BN-HER2 administra- tion was accompanied by increased tumor infiltration by HER-2-specific cytotoxic T lymphocytes (CTL). These tumor-specific CTL had characteristics similar to antiviral CTL, including strong expression of activation markers and co-expression of IFNγ and TNFα. Combination with CTLA-4 blockade significantly increased the magnitude of HER-2-specific T cell responses, with a higher propor- tion co-expressing TNFα and/or IL-2 with IFNγ. Further- more, in mice treated with MVA-BN-HER2 (alone or in combination with CTLA-4 blockade), the inducible T cell Electronic supplementary material The online version of this article (doi:10.1007/s00262-016-1816-7) contains supplementary material, which is available to authorized users. * Susan P. Foy [email protected] 1 Bavarian Nordic, Inc., 595 Penobscot Dr., Redwood City, CA 94063, USA 2 Present Address: ExoThera LLC, 675 Olive Street, Menlo Park, CA 94025, USA

Upload: others

Post on 25-Jul-2020

6 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Poxvirus-based active immunotherapy synergizes with CTLA-4 … · Cancer Immunol Immunother (2016) 65:537–549 DOI 10.1007/s00262-016-1816-7 ORIGINAL ARTICLE Poxvirus‑based active

1 3

Cancer Immunol Immunother (2016) 65:537–549DOI 10.1007/s00262-016-1816-7

ORIGINAL ARTICLE

Poxvirus‑based active immunotherapy synergizes with CTLA‑4 blockade to increase survival in a murine tumor model by improving the magnitude and quality of cytotoxic T cells

Susan P. Foy1 · Stefanie J. Mandl1 · Tracy dela Cruz1 · Joseph J. Cote1 · Evan J. Gordon1 · Erica Trent1 · Alain Delcayre1,2 · James Breitmeyer1 · Alex Franzusoff1 · Ryan B. Rountree1

Received: 5 November 2014 / Accepted: 23 February 2016 / Published online: 10 March 2016 © The Author(s) 2016. This article is published with open access at Springerlink.com

co-stimulator (ICOS) protein was expressed predominantly on CD4 and CD8 effector T cells but not on regulatory T cells (Treg). In contrast, mice left untreated or treated solely with CTLA-4 blockade harbored elevated ICOS+ Treg, a phenotype associated with highly suppressive activity. In conclusion, poxvirus-based active immunotherapy induced robust tumor infiltration by highly efficient effector T cells. Combination with CTLA-4 immune checkpoint block-ade amplified this response resulting in synergistically improved efficacy. These hypothesis-generating data may help elucidate evidence of enhanced clinical benefit from combining CTLA-4 blockade with poxvirus-based active immunotherapy.

Keywords Active immunotherapy · Poxvirus · Modified Vaccinia Ankara (MVA) · Cancer · Anti-CTLA-4 · Immune checkpoint blockade

AbbreviationsCEA Carcinoembryonic antigenCI Combination indexCTL Cytotoxic T lymphocytesCTLA-4 Cytotoxic T lymphocyte-associated

antigen-4CV-301 Poxvirus-based active immunotherapy

targeting CEA and MUC-1DPECs Double-positive effector cellsEECs Early effector cellsHER-2 Human epidermal growth factor

receptor 2HER2 Modified extracellular domain of

HER-2 and two additional T helper epitopes

ICOS Inducible T cell co-stimulatorIFN Interferon

Abstract The dramatic clinical benefit of immune check-point blockade for a fraction of cancer patients suggests the potential for further clinical benefit in a broader can-cer patient population by combining immune checkpoint inhibitors with active immunotherapies. The anti-tumor efficacy of MVA-BN-HER2 poxvirus-based active immu-notherapy alone or in combination with CTLA-4 check-point blockade was investigated in a therapeutic CT26-HER-2 lung metastasis mouse model. MVA-BN-HER2 immunotherapy significantly improved the median over-all survival compared to untreated controls or CTLA-4 blockade alone (p < 0.001). Robust synergistic efficacy was achieved with the combination therapy (p < 0.01). Improved survival following MVA-BN-HER2 administra-tion was accompanied by increased tumor infiltration by HER-2-specific cytotoxic T lymphocytes (CTL). These tumor-specific CTL had characteristics similar to antiviral CTL, including strong expression of activation markers and co-expression of IFNγ and TNFα. Combination with CTLA-4 blockade significantly increased the magnitude of HER-2-specific T cell responses, with a higher propor-tion co-expressing TNFα and/or IL-2 with IFNγ. Further-more, in mice treated with MVA-BN-HER2 (alone or in combination with CTLA-4 blockade), the inducible T cell

Electronic supplementary material The online version of this article (doi:10.1007/s00262-016-1816-7) contains supplementary material, which is available to authorized users.

* Susan P. Foy [email protected]

1 Bavarian Nordic, Inc., 595 Penobscot Dr., Redwood City, CA 94063, USA

2 Present Address: ExoThera LLC, 675 Olive Street, Menlo Park, CA 94025, USA

Page 2: Poxvirus-based active immunotherapy synergizes with CTLA-4 … · Cancer Immunol Immunother (2016) 65:537–549 DOI 10.1007/s00262-016-1816-7 ORIGINAL ARTICLE Poxvirus‑based active

538 Cancer Immunol Immunother (2016) 65:537–549

1 3

IL InterleukinKLRG1 Killer cell lectin-like receptor G1mCRPC Metastatic castration-resistant prostate

cancerMFI Mean fluorescence intensitymOS Median overall survivalMPECs Memory precursor effector cellsMUC-1 Mucin-1MVA Modified vaccinia AnkaraMVA-BN-HER2 Poxvirus-based active immunotherapy

targeting HER-2PAP Prostatic acid phosphatasePROSTVAC Poxvirus-based active immunotherapy

targeting PSAPSMA Prostate-specific membrane antigenPSA Prostate-specific antigenSLECs Short-lived effector cellsTeff Effector T cellsTILs Tumor-infiltrating lymphocytesTNF Tumor necrosis factorTregs Regulatory T cells

Introduction

The recent success of immune checkpoint inhibitors for the treatment of cancer has rejuvenated the field of cancer immunotherapy. Ipilimumab (Yervoy), a humanized mono-clonal antibody specific for cytotoxic T lymphocyte-associ-ated antigen-4 (CTLA-4), was the first FDA-approved drug from this new class of cancer immunotherapies. In patients with metastatic melanoma, ipilimumab treatment improved median overall survival (mOS) by 3.6 months and gener-ated a complete response rate with durable tumor regres-sion in 10–20 % of patients [1, 2]. Even greater clinical benefit may be achieved by selectively accelerating and augmenting anticancer immune responses through combi-nation with active cancer immunotherapy.

PROSTVAC, a poxvirus-based active immunotherapy targeting the prostate-specific antigen (PSA), resulted in an 8.5-month increase in the mOS (25.1 vs. 16.6 months, p = 0.0061) in a randomized, placebo-controlled Phase 2 clinical trial of men with asymptomatic or minimally symptomatic castration-resistant metastatic prostate cancer (mCRPC) [3]. In a similar patient population, a Phase 1 trial of 30 subjects combined PROSTVAC immunotherapy with dose escalation of ipilimumab (1, 3, 5 and 10 mg/kg per dose), which resulted in a mOS of 31.3 months for all groups combined [4–6]. Patients who received PROSTVAC plus 10 mg/kg ipilimumab had a mOS of 37.2 months, and there appeared to be a tail on the survival curve with 20 % of patients alive at 80 months [6]. In addition, 58 % of chemotherapy-naïve

subjects exhibited a PSA decline from baseline from the combination therapy [4].

Poxvirus-based active immunotherapies in additional cancer types are being investigated for clinical benefit as single-agent regimens, but clinical regimens exploring combinations with immune checkpoint inhibitors have not yet been initiated. For example, longer overall sur-vival following treatment with CV-301, a poxvirus-based active immunotherapy which targets carcinoembryonic antigen (CEA) and mucin-1 (MUC-1), was observed com-pared to contemporary controls in metastatic colon cancer (p < 0.0001) [7]. In addition, MVA-BN-HER2, a recom-binant modified vaccinia Ankara (MVA) vector express-ing a modified form of the extracellular domain of human epidermal growth factor receptor 2 (HER-2), is under development for the treatment of breast cancer. MVA-BN-HER2 was well tolerated in two Phase 1 clinical trials and showed evidence of peripheral immune responses against HER-2 in more than two-thirds of subjects [8]. Evidence that anti-tumor efficacy is mediated by induction of HER-2-specific tumor-infiltrating lymphocytes (TILs) was gen-erated in preclinical studies using MVA-BN-HER2 immu-notherapy [9].

In this report, the impact of combining MVA-BN-HER2 active immunotherapy and CTLA-4 blockade on efficacy was evaluated in a therapeutic mouse tumor model. We then investigated the immune response profiles of periph-eral and tumor-infiltrating T cells and identified character-istics that correlated with anti-tumor efficacy.

Materials and methods

Animals, tumor cell lines, and reagents

The CT26 murine colon carcinoma cell line express-ing human HER-2 (CT26-HER-2) was licensed from the Regents of the University of California [10]; the genetic profile matched the established CT26 cell line (Idexx Radil, Columbia, MO, USA). A master cell bank and working cell banks were generated, and each bank was tested positive for HER-2 expression by flow cytometry (data not shown). Each bank was tested for a comprehensive list of pathogens including mycoplasma by Idexx Radil and was pathogen free. Cells were maintained and used as previously pub-lished [9].

Female BALB/c mice (6–8 weeks old, Simonsen Lab-oratories, Gilroy, CA) were implanted i.v. with 5.0 × 104 CT26-HER-2 cells in 300 µL DPBS, which forms tumors in the lungs. On days 3 and 17 mice were treated with 200 µg of an antagonist anti-CTLA-4 antibody (Clone 9D9, Mouse IgG2b, Bio X Cell, West Lebanon, NH) by i.p. injection of 100 µL PBS. In studies to test for synergy and

Page 3: Poxvirus-based active immunotherapy synergizes with CTLA-4 … · Cancer Immunol Immunother (2016) 65:537–549 DOI 10.1007/s00262-016-1816-7 ORIGINAL ARTICLE Poxvirus‑based active

539Cancer Immunol Immunother (2016) 65:537–549

1 3

for the multicytokine functional assay, mice were treated with a dose titration of anti-CTLA-4 (200, 66, and 22 µg i.p. in 100 µL PBS) on days 3 and 17 or 4 and 18. On days 4 and 18 mice were treated with 1.0 × 107 Inf.U MVA-BN-HER2 in 7.1 µL by tail scarification (t.s.). MVA-BN-HER2 (Bavarian Nordic, BN, Martinsried, Germany) is a modified vaccinia Ankara-based recombinant vector that encodes a modified form of the human epidermal growth factor receptor 2 (HER-2), referred to as HER2 [9]. The modified HER2 comprises the extracellular domains of HER-2 and contains two additional T helper epitopes to enhance immunogenicity [11]. The infectious unit titer (Inf.U/mL) of MVA-BN-HER2 was determined by flow cytometry [12]. Control mice were implanted with CT26-HER-2 cells on day 1 and received no additional treatment. Six months after the primary CT26-HER-2 challenge, mice that rejected tumors were implanted with 5.0 × 104 CT26-WT cells in 300 µL DPBS.

To assess tumor burden, mice were euthanized on day 25 and lungs were weighed. In each group, 1–2 mice were euthanized and perfused with trypan blue (0.4 %, Medi-atech, Inc., Manassas, VA) through the trachea; the lungs were collected, briefly submerged in hydrogen perox-ide (30 %, EMD Millipore Corporation, Billerica, MA), washed in PBS, and photographed. For survival studies mice were euthanized at the first sign of distress. The BN Institutional Animal Care and Use Committee approved all animal procedures.

Flow cytometry

Mice were treated as described above, and whole blood was collected via tail vein on days 10/11, 24/25 or 38. A strong T cell response was measured at day 24/25, 1 week after the second virus treatment. Subsequent studies exam-ined tumor tissue at this time point when mice from each treatment as well as the control were alive. On day 24 or 25 whole blood was isolated by cardiac stick followed by col-lection of spleens and lungs for flow cytometric analysis. Tissues of 4–8 individual mice were analyzed for surface stains; tissues were pooled for 4–10 mice per group for the multicytokine and degranulation functional assays. Spleno-cytes were prepared by pressing the spleens between two frosted glass slides and lysing the red blood cells with ACK lysis buffer (Life Technologies, Grand Island, NY). Lungs and associated tumors were diced to ~1- to 2-mm3 pieces and further digested to single-cell suspensions for 1 h at 37 °C in DMEM with 10 % FBS, 50 U/mL DNAse I and 250 U/mL Collagenase I (Worthington Biochemical Corpo-ration, Lakewood, NJ). Red blood cells in lungs and whole blood were lysed with RBC lysis buffer (eBioscience).

Antibodies against the following proteins were pur-chased from BD Bioscience (San Jose, CA): CD3e

(500A2), CD4 (RM4-5), CD8a (53-6.7), CD11c (HL3), CD44 (IM7), CD62L (MEL-14), CD107a (1D4B), interferon-γ (IFN-γ) (XMG1.2), killer cell lectin-like receptor G1 (KLRG1, 2F1); from BioLegend (San Diego, CA): CD3e (145-2C11), CD127 (A7R34), IFNγ (XMG1.2), interleukin 2 (IL-2) (JES6-5H4), tumor necro-sis factor-α (TNFα) (MP6-XT22); and from eBiosci-ence (San Diego, CA): CD25 (PC61.5), ICOS (7E.17G9), CD127 (A7R34), FoxP3 (FJK-16 s), and CD16/CD32 (93).

To identify degranulating, cytotoxic T cells, single-cell suspensions of splenocytes (2 × 106 cells/well) or tumor/lungs (1 × 106 cells/well) were resuspended in RPMI-10 (10 % FBS, 1 % Pen-strep, and 0.1 % β-mercaptoethanol) and restimulated overnight at 37 °C in the presence of anti-CD107a antibody and Golgi Stop (BD Bioscience) [13]. The following peptides were synthesized by and pur-chased from JPT Peptide Technologies, Inc. (Acton, MA): MVA E3L and F2L (VGPSNSPTF and SPGAAGYDL, 1 µM each), HER-2 p63 (TYLPTNASL, 1 µM), HER-2 ECD overlapping peptide library (HER-2 OPL, 1 µM), and PSA (HPQKVTKFML, 1 µM) [9, 14–16]. Concanavalin A (ConA, MP Biomedicals, Santa Ana, CA) was used at 5 µg/mL as a positive control. The next day, cells were washed, blocked with anti-CD16/CD32 antibodies, and stained for surface markers. Cells were then washed, fixed/permeabi-lized with BD Cytofix/Cytoperm buffer, and stained intra-cellularly for IFNγ.

Additional intracellular cytokine staining was performed on splenocytes as described above except the anti-CD107a antibody was omitted, Golgi Stop and Golgi Plug were added, and cells were stained intracellularly for IFNγ, IL-2, and TNFα.

Regulatory T cells (Tregs) were stained intracellularly for FoxP3 using the fixation/permeabilization buffers from eBioscience according to the manufacturer’s instructions.

All samples were acquired on the BD LSRII or Fortessa and analyzed using FlowJo version 9.6.2 (TreeStar Inc., Ashland, OR). For all flow cytometry graphs, the CD4+ and CD8+ T cells were gated on CD3+ lymphocytes.

Statistical analysis

Statistical analyses were performed as described in the fig-ure legends using GraphPad Prism version 6.01 for Win-dows (GraphPad Software, La Jolla, CA). For immunolog-ical data, data shown are the mean and standard error of the mean (SEM). An analysis of variance (ANOVA) with Holm–Sidak multiple comparisons test was used to deter-mine statistical significance between treatment groups.

Synergy with the combination therapy in CT26-HER-2 challenged mice was determined by calculating the com-bination index (CI) using the Chou–Talalay method and CompuSyn software (www.combosyn.com) [17]. The

Page 4: Poxvirus-based active immunotherapy synergizes with CTLA-4 … · Cancer Immunol Immunother (2016) 65:537–549 DOI 10.1007/s00262-016-1816-7 ORIGINAL ARTICLE Poxvirus‑based active

540 Cancer Immunol Immunother (2016) 65:537–549

1 3

method requires performing a dose titration of each therapy alone and in combination. The effect at each dose is defined as the fractional survival (fa) and is based on the mOS for each dose. Hundred days was considered long-term sur-vival (fa ≈ 1). For example, with a mOS of 50 days, the fa = 0.5 (50 days/100 days). Multiple studies with the same treatment groups were pooled to determine the mOS. It was necessary to normalize the mOS with respect to the pooled control group to perform the analysis. CI < 1 indicates syn-ergism, CI = 1 indicates additive effect, and CI > 1 indi-cates antagonism.

Results

MVA‑BN‑HER2 combined with an anti‑CTLA‑4 antibody eliminates tumors in an experimental lung metastasis model

To test whether the efficacy of MVA-BN-HER2 immu-notherapy is augmented when combined with CTLA-4 immune checkpoint inhibition, mice were treated with MVA-BN-HER2 alone or in combination with an anti-CTLA-4 blocking antibody in a therapeutic HER-2+ exper-imental lung metastasis model. The combination of active immunotherapy plus checkpoint inhibition significantly and synergistically improved overall survival compared to either therapy alone [Fig. 1a, p < 0.001 vs. anti-CTLA-4 and p < 0.01 vs. MVA-BN-HER2; very strong synergy shown with a combination index (CI) < 0.001, see Sup-plemental Table 1]. The mOS of the combination therapy was greater than 100 days, significantly longer than either MVA-BN-HER2 treatment (49.5 days) or anti-CTLA-4 treatment (35 days) alone. In addition, more than half of the mice treated with the combination therapy (57 %) were still alive 100 days after tumor challenge, compared to 14 % of mice treated with MVA-BN-HER2 alone or 8 % of mice treated with anti-CTLA-4 alone. Next we tested the longevity of the induced anti-tumor response

and evaluated whether the response had spread to antigens not encoded by the poxvirus-based active immunotherapy (antigen spreading). A small subset of mice that success-fully rejected the initial CT26-HER-2 lung metastasis were rechallenged with CT26-WT cells (not expressing HER-2)

Days post tumor challenge

Perc

ent s

urvi

val

0 20 40 60 80 1000

50

100

Control MVA-BN-HER2MVA-BN-HER2 + anti-CTLA-4anti-CTLA-4

2/25

4/28

16/28

** ***

Naive

Control

anti-C

TLA-4

MVA-BN-H

ER2

MVA-BN-H

ER2

+ anti-C

TLA-4

0.0

0.2

0.4

Lung

wei

ght (

g)

0.6

0.8

1.0

****

MVA-BN-HER2MVA-BN-HER2+ anti-CTLA-4

Naïve Control anti-CTLA-4

(A)

(B)

(C)

Fig. 1 Overall survival and anti-tumor efficacy with MVA-BN-HER2 immunotherapy and CTLA-4 blockade. a Kaplan–Meier sur-vival curves from the combined results of three independent studies. The median overall survival in control mice (30 days) was signifi-cantly extended with anti-CTLA-4 treatment (35 days, **p < 0.01), MVA-BN-HER2 treatment (49.5 days, ***p < 0.001), and MVA-BN-HER2+ anti-CTLA-4 combination therapy (>100 days, ***p < 0.001). A log-rank test with Bonferroni correction was used to determine significance between groups. b Representative lungs from each group harvested at day 25 were stained with Trypan Blue. Healthy lung tissue appears blue, while tumors appear red and white. Scale bar 1 cm. c Lung weights from treated mice on day 25 com-pared to naïve mice. A one-way ANOVA with Dunnett’s multiple comparisons test was used to determine significance between groups, ****p < 0.0001

Page 5: Poxvirus-based active immunotherapy synergizes with CTLA-4 … · Cancer Immunol Immunother (2016) 65:537–549 DOI 10.1007/s00262-016-1816-7 ORIGINAL ARTICLE Poxvirus‑based active

541Cancer Immunol Immunother (2016) 65:537–549

1 3

6 months after primary challenge. All rechallenged mice rejected the parental tumors, while naïve mice challenged with the same parental tumor cells had a mOS of 26 days (Supplemental Figure 1). These data demonstrate that long-lasting immune responses and antigen spread (expansion of the T cell repertoire from HER-2 to other cancer antigens) had occurred and were potent enough to protect mice from challenge with non-HER-2-expressing tumors.

Lungs of identically treated mice were removed 25 days after tumor implantation and assessed for tumor burden by lung weight. As seen in Fig. 1b, c, lungs of control mice or anti-CTLA-4 treated mice had numerous visible tumors at day 25, and the average lung weight was more than dou-bled compared to healthy naïve mice (p < 0.0001). In stark contrast, lungs of mice treated with MVA-BN-HER2 alone or in combination with anti-CTLA-4 appeared healthy with a similar appearance as the lungs of naïve unchallenged mice, and had no difference in lung weight (p > 0.99). These results highlight the benefit of MVA-BN-HER2 treatment to increase survival and reduce tumor burden.

Poxvirus‑based immunotherapy results in a distinct effector T cell phenotype

We hypothesized that phenotypic and functional analy-sis of the effector T cell responses at the predicted peak

of treatment-induced activity, 1 week after the last treat-ment cycle, would reveal differences that later manifest as improved overall survival. Therefore, phenotypes of peripheral (spleen and blood) and tumor-/lung-infiltrating T cells from each treatment cohort were evaluated on day 25 by flow cytometry.

To determine whether there was a specific phenotype associated with MVA-BN-HER2 treatment that was aug-mented in combination with CTLA-4 blockade, we first looked at memory and activation markers induced by the different treatments. Effector memory cells generated in response to virus stimulation can be classified based on their expression of CD127 (IL7-Rα) and KLRG1. The KLRG1− CD127+ subset is a long-lived memory precursor effector cell (MPEC) population, while the KLRG1+ CD127− sub-set is known as short-lived effector cells (SLECs) [18]. The KLRG1+ CD127+ double-positive effector cells (DPECs) are believed to constitute a potent long-lived effector memory T cell subset [18, 19]. SLEC and DPEC CD8 T cell populations were detected in the lungs and peripheral tissue of mice following MVA-BN-HER2 administration, but not after anti-CTLA-4 treatment (Fig. 2a, b). Further analysis showed that the vast majority (>80 %) of both SLECS and DPECS were CD44hi CD62L− (Supplemental Figure 2). The quality of the response induced by poxvirus-based immunotherapy was maintained by combination with

Naïve Control anti-CTLA-4 MVA-BN-HER2 MVA-BN-HER2 + anti-CTLA-4

MPECs

EECs SLECs

DPECsK

LRG

1+ C

D12

7- SLE

Cs

% o

f CD

8+ T

Cel

ls

Tumor/Lungs Spleen Blood Tumor/Lungs Spleen Blood0

10

20

30

*†*†

*†*†

*†*†

KLR

G1+

CD

127+

DPE

Cs

% o

f CD

8+ T

Cel

ls

0

10

20

30

*† *†

*†*† *†

*†

KLRG1CD

127

KLRG1

CD12

7anti-CTLA-4ControlNaïve MVA-BN-HER2

MVA-BN-HER2 + anti-CTLA-4

0.1 0.2 0.3 15.3 29.9

2.7 3.0 3.6 30.4 33.2

61.7 54.3 53.7 37.9 28.0

(A)

(B) (C)

Fig. 2 Effector T cell populations after MVA-BN-HER2 immuno-therapy and/or CTLA-4 blockade. a Schematic for characterization of cells and a representative example of CD127 and KLRG1 expres-sion in the tumor/lungs at day 25. b CD8+ CD127− KLRG1+ short-lived effector cells (SLECs) and c CD8+ CD127+ KLRG1+ double-

positive effector cells (DPECs). MPECS memory precursor effector cells, EECs early effector cells. n = 8 mice/group combined from two independent experiments. p < 0.05: * versus control, † versus anti-CTLA-4, and # versus MVA-BN-HER2

Page 6: Poxvirus-based active immunotherapy synergizes with CTLA-4 … · Cancer Immunol Immunother (2016) 65:537–549 DOI 10.1007/s00262-016-1816-7 ORIGINAL ARTICLE Poxvirus‑based active

542 Cancer Immunol Immunother (2016) 65:537–549

1 3

anti-CTLA-4, but it was not augmented by combination therapy. Hence, the characterization of these SLEC and DPEC T cell populations at this time point was valuable for monitoring productive immunity in response to poxvirus-mediated immunotherapy.

Combination therapy enhanced the population of tumor antigen‑specific cytotoxic TILs

To determine whether the improved survival could be due to enhanced CTL function, antigen-specific cytotoxic T cell activity in the tumor/lungs and the periphery (spleen) was evaluated. MVA-BN-HER2 immunotherapy plus CTLA-4 checkpoint blockade led to a marked increase in the proportion of functional, HER-2-specific CD8 cyto-toxic T cells infiltrating into tumor tissue (Fig. 3a). While MVA-BN-HER2 or anti-CTLA-4 therapy alone resulted in

moderate induction of HER-2-specific CD8 TILs, there was no response in control mice. Of note, the HER-2-specific cytotoxic CD8 response was three- to fourfold higher in the tumor/lungs than in the spleen, while the virus-targeted response (i.e., stimulated by MVA-specific E3L and F2L peptides) alone or in combination with anti-CTLA-4 was similar in both tissues. Thus, HER-2-specific T cells pref-erentially homed to the tumor, and the magnitude of HER-2-specific CD8 TILs response correlated with the length of survival in the tumor model.

The degranulating cells that responded to either HER-2 p63 or MVA restimulation were predominantly SLECs (Fig. 3b), suggesting that the effector memory functions associated with the viral response phenotype also charac-terized cells responding to the HER-2 p63 antigen. Over-all, anti-CTLA-4 monotherapy increased the cytotoxic CD8 TILs tenfold compared to mice that had received no

HER-2 Specific ResponseMVA Specific Response

Control200

anti-CTLA-4100

MVA-BN-HER211300

MVA-BN-HER2+ anti-CTLA-4

24200

Control800

anti-CTLA-41600

MVA-BN-HER213500

MVA-BN-HER2+ anti-CTLA-4

17500

Control100

anti-CTLA-4200

MVA-BN-HER21900

MVA-BN-HER2+ anti-CTLA-4

4300

Control200

anti-CTLA-42300

MVA-BN-HER25300

MVA-BN-HER2+ anti-CTLA-4

14900

Spleen

Tumor/Lungs

Spleen

Tumor/Lungs

KLRG1+ CD127+ DPECsKLRG1+ CD127- SLECsKLRG1- CD127+ MPECsKLRG1- CD127- EECs

0

2

4

6

8

10C

D10

7+ IFNγ+

(% o

f CD

8+ T c

ells

)

Tumor/Lungs

Spleen

MVA E3L F2L Media (-) ConA (+)HER-2 p63 PSA (-)

--

+-

++

-+

MVA-BN-HER2anti-CTLA-4

--

+-

++

-+

--

+-

++

-+

--

+-

++

-+

--

+-

++

-+

(A)

(B)

Fig. 3 Degranulating T cells in the tumor/lungs or spleen of mice treated with MVA-BN-HER2 and/or CTLA-4 blockade. a Virus (MVA E3L F2L) and tumor antigen (HER-2 p63) specific responses were measured in the tumor/lungs and spleen; tissue was pooled from

4 mice/group. b Expression of KLRG1 and CD127 on the virus or HER-2 p63-degranulating (CD107a+ IFNγ+) cells from A. Pie charts are area-weighted to reflect the number of CD8+ CD107a+ IFNγ+ cells per million T cells

Page 7: Poxvirus-based active immunotherapy synergizes with CTLA-4 … · Cancer Immunol Immunother (2016) 65:537–549 DOI 10.1007/s00262-016-1816-7 ORIGINAL ARTICLE Poxvirus‑based active

543Cancer Immunol Immunother (2016) 65:537–549

1 3

treatment. However, MVA-BN-HER2 administration led to a 25-fold increase in numbers of HER-2-specific cytotoxic CD8 TILs compared to no treatment. This impact on HER-2-specific cytotoxic CD8 TILs was augmented to a 75-fold increase over controls following combination of active MVA-BN-HER2 immunotherapy with CTLA-4 checkpoint blockade.

Combination therapy induces the expansion of polyfunctional CD8 T cells

The quality of the T cell response was further character-ized by measuring IFNγ, TNFα, and IL-2 cytokine levels in stimulated splenic CD8+ T cells. In response to virus or

HER2-p63 restimulation, a five- to tenfold increase in the magnitude of IFNγ+ T cells was found in mice treated with MVA-BN-HER2 compared to tumor-bearing mice that received no treatment (control) or CTLA-4 blockade alone, as shown by the relative size of the pie charts (Fig. 4a). The magnitude of the response to combination treatment was on average twofold larger as compared to the MVA-BN-HER2 treatment group after HER2-p63 restimulation (p < 0.01). The significant increase in IFNγ production with the com-bination therapy compared to MVA-BN-HER2 alone was observed only when splenocytes were restimulated with the tumor-specific antigen (HER2-p63) and not in response to restimulation with the poxvirus (MVA). Following MVA-BN-HER2 treatment, the expansion of IFNγ-producing

MVA Specific Response

Control300

anti-CTLA-4700

MVA-BN-HER27500

MVA-BN-HER2+ anti-CTLA-4

13700

HER-2 Specific Response

Control300

anti-CTLA-4500

MVA-BN-HER22000

MVA-BN-HER2+ anti-CTLA-4

5300

IFNγ+ TNFα+ IL-2+

IFNγ+ TNFα+

IFNγ+ IL-2+

IFNγ+

0

5,000

10,000

15,000

20,000

IFN

γ MFI

IFN

γ MFI

--

+-

++

-+

MVA-BN-HER2anti-CTLA-4

0

5,000

10,000

15,000

--

+-

++

-+

MVA-BN-HER2anti-CTLA-4

0

100

200

300

IFN

γ+ T

NFα

+ IL

-2+

CD

8+ /1

E6 T

cel

ls

MVA-BN-HER2anti-CTLA-4

--

+-

++

-+

#* †

0

1000

2000

3000

4000

5000

IFN

γ+ T

NFα

+C

D8+

/1E6

T c

ells

MVA-BN-HER2anti-CTLA-4

--

+-

++

-+

#* †

0

100

200

300

400

IFN

γ+ IL

-2+

CD

8+ /1

E6 T

cel

ls

MVA-BN-HER2anti-CTLA-4

--

+-

++

-+

0

1000

2000

3000

IFN

γ+C

D8+

/1E6

T c

ells

MVA-BN-HER2anti-CTLA-4

--

+-

++

-+

*†

0

500

1000

1500

2000

2500

IFN

γ+ T

NFα

+ IL

-2+

CD

8+ /1

E6 T

cel

ls

--

+-

++

-+

MVA-BN-HER2anti-CTLA-4

*†

0

5000

10000

15000

IFN

γ+ T

NFα

+C

D8+

/1E6

T c

ells

MVA-BN-HER2anti-CTLA-4

--

+-

++

-+

* †

0

500

1000

1500

2000

IFN

γ+ IL

-2+

CD

8+ /1

E6 T

cel

ls #

MVA-BN-HER2anti-CTLA-4

--

+-

++

-+

* †

* †

0

5000

10000

15000

IFN

γ+C

D8+

/1E6

T c

ells

MVA-BN-HER2anti-CTLA-4

--

+-

++

-+

* †* †

(A)

(B)

(C)

Fig. 4 Antigen-specific cytokine production in CD8 T cells in the spleen. a Cytokine production in the spleen of mice treated with MVA-BN-HER2 and/or CTLA-4 blockade. Pie charts are area-weighted to reflect the number of CD8+ IFNγ+ cells per million T cells. b IFNγ, TNFα, and IL-2 subsets of cytokine+ T cells shown in

a. c IFNγ MFI plotted for the subsets of cytokine+ T cells shown in A. Data combined from 5 experiments. In each experiment, spleno-cytes of 4–10 mice/group were pooled. p < 0.05: * versus control, † versus anti-CTLA-4, and # versus MVA-BN-HER2

Page 8: Poxvirus-based active immunotherapy synergizes with CTLA-4 … · Cancer Immunol Immunother (2016) 65:537–549 DOI 10.1007/s00262-016-1816-7 ORIGINAL ARTICLE Poxvirus‑based active

544 Cancer Immunol Immunother (2016) 65:537–549

1 3

antigen-specific cells was accompanied by a shift to a polyfunctional cytokine phenotype. For instance, CTLA-4 blockade alone induced CD8 T cells that were predomi-nantly IFNγ single positive cells (depicted in purple). In contrast, more than 50 % of the IFNγ positive cells in MVA-BN-HER2-treated animals also produced TNFα (depicted in green) or IL-2 (depicted in blue), and a sub-set of those cells produced all three cytokines (depicted in orange). Combination treatment resulted in a statistically significant increase in this proportion of tumor antigen-specific (HER2-p63) cytokine-producing effector cells (Fig. 4b). A significantly higher percentage of the IFNγ+ TNFα+ IL-2+ or IFNγ+ TNFα+ polyfunctional HER-2 spe-cific T cells were observed for the combination therapy as compared to MVA-BN-HER2 alone. This increase was spe-cific for the HER-2 tumor antigen and was not observed in response to poxvirus-specific restimulation (MVA). Exami-nation of the levels of IFNγ production from each of these CD8 T cell subsets was quantified by the mean fluorescence intensity (MFI) of each functional phenotype (Fig. 4c). On a per cell basis, polyfunctional cells produced more IFNγ than single positive cells. Overall, the cytokine profiles indicate that the functional quality of the tumor antigen-specific T cell response, in addition to the magnitude of the tumor-specific T cell response, is augmented even further by the combination of active immunotherapy plus CTLA-4 checkpoint blockade.

Poxvirus‑based active immunotherapy results in markedly improved Teff to Treg ratios within the tumor microenvironment

The expression profiles of the inducible T cell co-stimulatory (ICOS) molecule were of particular interest because this T cell activation marker is known to increase on CD4+ T cells within the tumor and periphery following anti-CTLA-4 treat-ment [20]. CD4+ T cells exhibited elevated ICOS expression in the tumor/lungs of untreated controls and all treatment groups compared to naïve, tumor-free mice (Fig. 5a, b). Therefore, ICOS expression was induced on CD4+ T cells by the presence of the tumor without any treatment. Addition of the anti-CTLA-4 antibody alone raised ICOS expression to an even higher percentage of CD4+ T cells. Interestingly, ICOS levels were somewhat reduced on CD4+ T cells fol-lowing MVA-BN-HER2 treatment compared to untreated controls, but were again higher when MVA-BN-HER2 was combined with CTLA-4 blockade. The increase in ICOS on CD4+ T cells was most prominent within the tumor/lungs, but elevated levels were also detected in the periphery of the treated groups but not in the control, tumor-bearing mice. Therefore, although differences in ICOS expression on CD4+ T cells were observed, they were not alone character-istic of protective anti-tumor immune responses.

Analysis of CD8 T cells revealed that in the tumor/lungs ICOS expression showed the highest increases in MVA-BN-HER2-treated mice irrespective of CTLA-4 treatment (Fig. 5c). In the blood and spleen, ICOS expression was consistently elevated on CD8+ T cells of mice treated with MVA-BN-HER2 in combination with anti-CTLA-4, com-pared to control or anti-CTLA-4 treated mice. Thus, while the percentage of ICOS+ CD8+ T cells was significantly higher in the periphery with combination treatment, the responses observed in the tumor were similar in MVA-BN-HER-2-treated groups irrespective of anti-CTLA-4 treat-ment. This demonstrates that the poxvirus-based immuno-therapy drives the quality of the induced immune response.

Elevated ICOS expression on CD4+ T cells characterizes the activation of effector cells (Teff) as well as highly sup-pressive Tregs [21, 22]. We therefore examined which class of CD4+ T cells expressed ICOS in the different treatment groups (Fig. 6a). ICOS was elevated on FoxP3− Teff cells but not Tregs following MVA-BN-HER2 treatment (Fig. 6b, c), and ICOS was even more pronounced on FoxP3− Teff cells following combination therapy (Fig. 6c). This resulted in a marked increase in the ratio of both CD4+ and CD8+ ICOS+ Teff to ICOS+ Tregs in the tumor/lungs and periphery in mice receiving MVA-BN-HER2 treatment compared to control mice (Fig. 6d, e). In contrast, in tumor-bearing con-trol mice and mice treated only with anti-CTLA-4 where tumor burden was high, ICOS expression was increased on both FoxP3+ Tregs and FoxP3− Teff cells (Fig. 6b, c). Overall, the high ICOS+ Teff to ICOS+ Treg ratio induced by MVA-BN-HER2 alone or in combination with CTLA-4 blockade likely reflects a more effective immune response and correlated with strong anti-tumor efficacy.

Discussion

We found that combining the poxvirus-based active immu-notherapy with CTLA-4 checkpoint blockade strongly syn-ergized to increase overall survival in a therapeutic mouse tumor model. In this combination, the poxvirus-based active immunotherapy drove the quality of the response, which was characterized by highly activated polyfunctional T cells; this T cell phenotype was further amplified by CTLA-4 treatment.

Treatment with MVA-BN-HER2, but not anti-CTLA-4 alone, led to a dramatic increase of KLRG1 expression on CD8 T cells which were categorized with CD127/IL-7Rα expression as short-lived effector cells (SLECs, KLRG1+ CD127−) or an effector-like memory subset of double-positive effector cells (DPECs, KLRG1+ CD127+) [19]. Induction of SLECs and DPECs was driven by the poxvi-rus-based immunotherapy and was not further expanded in the combination therapy group. The SLEC and DPEC

Page 9: Poxvirus-based active immunotherapy synergizes with CTLA-4 … · Cancer Immunol Immunother (2016) 65:537–549 DOI 10.1007/s00262-016-1816-7 ORIGINAL ARTICLE Poxvirus‑based active

545Cancer Immunol Immunother (2016) 65:537–549

1 3

phenotypes were also identified in mice treated with PROS-TVAC and MVA poxvirus-based active immunotherapies [23, 24]. Interestingly, the SLEC and DPEC populations described here expanded for both virus-specific and HER-2-specific degranulating T cells. This indicates that the effector characteristics associated with the viral response also defined the T cells responding to the encoded HER-2 tumor-associated antigen. It may therefore be possible to identify poxvirus-induced T cells responding to the inserted tumor antigens in patients treated with poxvirus-based active immunotherapy by including virally induced mark-ers in phenotyping panels [25].

The quality of the T cell responses is often assessed by the profile of cytokines produced upon activation,

with co-expression of multiple cytokines defining more active effector cells [26]. Examination of a multicytokine profile of IFNγ, TNFα, and IL-2 revealed that poxvirus immunotherapy promoted co-expression of cytokines on CD8 T cells. This expansion in polyfunctional T cells was present in response to both the virus and the tumor antigen. Combination treatment with CTLA-4 blockade significantly increased the number of IFNγ+ TNFα+ IL-2+ triple-positive and IFNγ+ TNFα+ double-positive cells specific for the HER-2 tumor antigen but not the poxvirus vector (MVA), and this correlated with increased mOS. Furthermore, the magnitude of HER-2-specific cytotoxic activity was increased in TILs of combination-treated animals.

Naïve Control anti-CTLA-4 MVA-BN-HER2 MVA-BN-HER2 + anti-CTLA-4

ICO

S+

% o

f CD

4+ T C

ells

ICO

S+

% o

f CD

8+ T C

ells

Tumor/Lungs Spleen Blood Tumor/Lungs Spleen Blood0

20

40

60

*

†# *

*

#

*

*

†* * #

*

0

10

20

30

40

50

* *

*† *†

*

ICOS

CD4

ICOS

CD8

anti-CTLA-4ControlNaïve MVA-BN-HER2MVA-BN-HER2 + anti-CTLA-4

10.6 40.8 53.4 23.6 38.3

3.2 15.4 21.3 24.5 26.4

(A)

(B) (C)

Fig. 5 ICOS expression on CD4+ and CD8+ T cells. a Representa-tive example of ICOS expression on CD4+ and CD8+ T cells in the tumor/lungs at day 25. b ICOS expression on CD4+ T cells in the tumor/lungs, spleen, and blood. c ICOS expression on CD8+ T cells.

n = 8 mice/group combined from two independent experiments. p < 0.05: * versus control, † versus anti-CTLA-4, and # versus MVA-BN-HER2

Page 10: Poxvirus-based active immunotherapy synergizes with CTLA-4 … · Cancer Immunol Immunother (2016) 65:537–549 DOI 10.1007/s00262-016-1816-7 ORIGINAL ARTICLE Poxvirus‑based active

546 Cancer Immunol Immunother (2016) 65:537–549

1 3

ICOS is expressed on activated T cells as a positive co-stimulatory molecule, and its expression on peripheral T cells may provide a biomarker for therapeutic benefit with anti-CTLA-4 therapy [20, 27–29]. Our results revealed that treatment with CTLA-4 blockade augmented the num-ber of ICOS+ CD4+ T cells but not ICOS+ CD8+ T cells in the tumor, blood, and spleen. In contrast, MVA-BN-HER2 treatment was generally accompanied by an expan-sion of ICOS+ CD8+ T cells, but this population was not further increased with CTLA-4 blockade. Though ICOS expression alone did not reveal the additional benefit of

combining poxvirus immunotherapy with checkpoint inhi-bition, these data suggest that higher expression of ICOS on effector cell populations may correlate with improved survival.

Importantly, poxvirus-based immunotherapy, alone or in combination with anti-CTLA-4 blockade, led to an increase in the ICOS+ Teff to ICOS+ Treg ratio. This shift in the ICOS+ Teff to ICOS+ Treg ratio was most pronounced at the tumor site, but it was also apparent in the spleen and blood. Unlike earlier reports, the Teff/Treg ratio did not increase following CTLA-4 inhibition alone [30–32], even though

FoxP

3+ ICO

S+

% o

f CD

4+ T C

ells

Tumor/Lungs Spleen Blood0

10

20

30

40

50

*

*† * #* *#

† †*

FoxP

3- ICO

S+

% o

f CD

4+ T C

ells

Tumor/Lungs Spleen Blood

Tumor/Lungs Spleen Blood Tumor/Lungs Spleen Blood

0

10

20

30

40

#

* *

#*

*

*#*

*

CD

4+ Tef

f:Tre

g

ICO

S+ FoxP

3- /ICO

S+ FoxP

3+

0

2

4

6

*†*†

**†

*†*†

CD

8+ Tef

f:Tre

g

CD

8+ ICO

S+ /CD

4+ ICO

S+ FoxP

3+

0

5

10

15

†*†*

†*

†*

ICOS

FoxP

3

Naïve Control anti-CTLA-4 MVA-BN-HER2 MVA-BN-HER2 + anti-CTLA-4

anti-CTLA-4ControlNaïve MVA-BN-HER2MVA-BN-HER2 + anti-CTLA-4

3.9 22.6 26.2 4.1 6.9

7.8 19.4 28.8 20.8 26.9

6.5 5.1 4.9 5.0 5.9

(A)

(B) (C)

(D) (E)

Fig. 6 ICOS expression on effector and regulatory T cells. a Repre-sentative example of ICOS and FoxP3 expression in the tumor/lungs at day 25. b ICOS expression on FoxP3+ Tregs. c ICOS expression on FoxP3− CD4 T cells. d CD4 ICOS+ Teff to ICOS+ Treg ratio (CD4+

ICOS+ FoxP3−/CD4+ ICOS+ FoxP3+) and e CD8 ICOS+ Teff to ICOS+ Treg ratio (CD8+ ICOS+/CD4+ ICOS+ FoxP3+). n = 8 mice/group combined from two independent experiments. p < 0.05: * ver-sus control, † versus anti-CTLA-4, and # versus MVA-BN-HER2

Page 11: Poxvirus-based active immunotherapy synergizes with CTLA-4 … · Cancer Immunol Immunother (2016) 65:537–549 DOI 10.1007/s00262-016-1816-7 ORIGINAL ARTICLE Poxvirus‑based active

547Cancer Immunol Immunother (2016) 65:537–549

1 3

there was a modest increase in overall survival (5 days). This may be due to a couple of factors. First, when the ICOS+ Teff to ICOS+ Treg ratio was measured mice receiv-ing CTLA-4 blockade alone had a large tumor burden, thus the shift in ICOS+ Teff to ICOS+ Treg populations may no longer have been apparent. Second, the isotype of the anti-CTLA-4 antibody is critical for the Treg depleting activity of the anti-CTLA-4 antibody [33]. The IgG2a isotype, but not the IgG2b isotype of the anti-CTLA-4 antibody used in the present study, led to selective reduction of intratumoral Tregs [33]. Previous studies have shown that combining MVA-BN-HER2 with Treg depletion further increased the mOS compared to MVA-BN-HER2 alone [9]. Therefore, combining MVA-BN-HER2 with an anti-CTLA-4 antibody with Treg depleting properties may lead to an even higher ICOS+ Teff to ICOS+ Treg ratio and could potentially further enhance the efficacy of the combination therapy.

Antigen spread is the development of novel immune responses against target antigens expressed by the patient’s own tumor. Antigen spread responses are thought to be a crucial component of successful immunotherapy as they expand the anticancer immune repertoire beyond the ini-tially targeted antigen. Rechallenge experiments with non-HER-2-expressing tumors demonstrated that the immune responses were durable and were accompanied by antigen spread to additional tumor antigens. These data corrobo-rate previous preclinical findings targeting HER-2, PSA, or CEA [9, 23, 34]. More importantly, antigen spread responses were also detected in clinical studies in 68 % of patients treated with PROSTVAC [35]. Immune responses to additional tumor antigens included PSMA (prostate-spe-cific membrane antigen), PAP (prostatic acid phosphatase), MUC-1, and Brachyury [4, 36].

The present study was performed in mice where human HER-2 is not a self-antigen, and immune responses could be lower in a tolerant model. Anti-tumor efficacy and immune responses were demonstrated in transgenic mice expressing human CEA as a self-antigen using CEA-targeted poxvirus-based active immunotherapy [37, 38]. Furthermore, when these poxvirus-based immunother-apy candidates were used in combination with CTLA-4 blockade, increased lymphocyte proliferation, IFNγ pro-duction, and T cell avidity resulted in tumor rejection through immune responses to self-antigens in tolerant mouse models [39–41]. Our present study further charac-terized the effect of combination therapy by investigating the magnitude and quality of TILs and peripheral lympho-cytes. Our examination of viral and tumor antigen-specific responses revealed that the effector T cell characteristics associated with the viral response also defined the T cells responding to the encoded HER-2 tumor antigen and were further augmented in combination with CTLA-4 block-ade. Thus, the preclinical data generated here provide

hypothesis-generating evidence for elucidating the poten-tial correlates of improved T cell effector responses result-ing from combination poxvirus-based immunotherapy plus CTLA-4 checkpoint blockade.

In the clinic, monotherapy with PROSTVAC, a PSA-targeted poxvirus-based active immunotherapy, signifi-cantly extended the mOS from 16.6 to 25.1 months in patients with metastatic castration-resistant prostate can-cer [3]. Ipilimumab alone provided a nonsignificant 1.2-month improvement in overall survival compared to the placebo group in a Phase 3 trial of patients with prostate cancer [42]. Combining PROSTVAC with CTLA-4 block-ade in a similar patient population resulted in an encourag-ing mOS of 31.3 months in a Phase 1 study of fixed dose PROSTVAC with a dose escalation of Ipilimumab [3, 5, 6]. Of note, 20 % of patients treated with the highest dose of ipilimumab were still alive at 80 months [6]. Importantly, immune-related adverse events commonly associated with immune checkpoint inhibition were not exacerbated by combination with PROSTVAC immunotherapy [4]. Thus, the data from this work elucidating potential immunologic correlates for improved anti-tumor response in preclini-cal models together with a suggestion of improved over-all survival in the clinic support further clinical investiga-tion of combination poxvirus-based active immunotherapy together with CTLA-4 immune checkpoint inhibition in prostate cancer and other cancers.

Conclusion

In summary, we showed that combination of poxvirus-based active immunotherapy with CTLA-4 blockade resulted in synergistic anti-tumor efficacy. Functional and phenotypic characteristics of the immune response were driven by the poxvirus-based active immunotherapy and the antigen-specific effector T cell responses were further amplified by CTLA-4 blockade. These data provide evi-dence that may translate into the potential immunologic correlates of productive anticancer immunity and clinical benefit resulting from poxvirus-based active immunother-apy combined with CTLA-4 immune checkpoint inhibition.

Acknowledgments Special thanks to Natasha Dubinina and Fareed Yahya for their expert animal handling and assistance in husbandry. We would like to thank Don Bellgrau, Nick Gaspar, Thierry Giffon, Henning Lauterbach, Rachel Owen, Dennis Panicali, and Titus Plattel for their critical review of the manuscript.

Authors’ contributions S.P. Foy, S.J. Mandl, A. Delcayre, and R.B. Rountree contributed to conception and design. S.P. Foy and T. dela Cruz were involved in the development of methodology. S.P. Foy, T. dela Cruz, J.J. Cote, E.J. Gordon, and E. Trent contributed to the acquisition of data. S.P. Foy, S.J. Mandl, T. dela Cruz, J.J. Cote, E.J. Gordon, E. Trent, A. Delcayre, A. Franzusoff, and R.B. Rountree

Page 12: Poxvirus-based active immunotherapy synergizes with CTLA-4 … · Cancer Immunol Immunother (2016) 65:537–549 DOI 10.1007/s00262-016-1816-7 ORIGINAL ARTICLE Poxvirus‑based active

548 Cancer Immunol Immunother (2016) 65:537–549

1 3

analyzed and interpreted the data. S.P. Foy, S.J. Mandl, T. dela Cruz, A. Delcayre, J. Breitmeyer, A. Franzusoff, and R.B. Rountree were involved in the writing, review, and/or revision of the manuscript.

Compliance with ethical standards

Conflicts of interest Research was conducted at and funded by Bavarian Nordic, Inc. All authors are current or former employees of Bavarian Nordic, Inc.

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://crea-tivecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

References

1. Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, Akerley W, van den Eertwegh AJ, Lutzky J, Lorigan P, Vaubel JM, Linette GP, Hogg D, Ottensmeier CH, Lebbe C, Peschel C, Quirt I, Clark JI, Wolchok JD, Weber JS, Tian J, Yellin MJ, Nichol GM, Hoos A, Urba WJ (2010) Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 363:711–723

2. Prieto PA, Yang JC, Sherry RM, Hughes MS, Kammula US, White DE, Levy CL, Rosenberg SA, Phan GQ (2012) CTLA-4 blockade with ipilimumab: long-term follow-up of 177 patients with metastatic melanoma. Clin Cancer Res 18:2039–2047

3. Kantoff PW, Schuetz TJ, Blumenstein BA, Glode LM, Bilhartz DL, Wyand M, Manson K, Panicali DL, Laus R, Schlom J, Dahut WL, Arlen PM, Gulley JL, Godfrey WR (2010) Overall survival analysis of a phase II randomized controlled trial of a Poxviral-based PSA-targeted immunotherapy in metastatic cas-tration-resistant prostate cancer. J Clin Oncol 28:1099–1105

4. Madan RA, Mohebtash M, Arlen PM, Vergati M, Rauckhorst M, Steinberg SM, Tsang KY, Poole DJ, Parnes HL, Wright JJ, Dahut WL, Schlom J, Gulley JL (2012) Ipilimumab and a poxviral vac-cine targeting prostate-specific antigen in metastatic castration-resistant prostate cancer: a phase 1 dose-escalation trial. Lancet Oncol 13:501–508

5. Jochems C, Tucker JA, Tsang KY, Madan RA, Dahut WL, Liewehr DJ, Steinberg SM, Gulley JL, Schlom J (2014) A com-bination trial of vaccine plus ipilimumab in metastatic castration-resistant prostate cancer patients: immune correlates. Cancer Immunol Immunother 63:407–418

6. Singh H, Madan RA, Dahut WL, O’Sullivan Coyne G, Rauck-horst M, McMahon S, Heery C, Schlom J, Gulley J (2015) Com-bining active immunotherapy and immune checkpoint inhibitors in prostate cancer. J Clin Oncol 33 (suppl 7; abstr 172)

7. Morse MA, Niedzwiecki D, Marshall JL, Garrett C, Chang DZ, Aklilu M, Crocenzi TS, Cole DJ, Dessureault S, Hobeika AC, Osada T, Onaitis M, Clary BM, Hsu D, Devi GR, Bulusu A, Annechiarico RP, Chadaram V, Clay TM, Lyerly HK (2013) A randomized phase II study of immunization with dendritic cells modified with poxvectors encoding CEA and MUC1 compared with the same poxvectors plus GM-CSF for resected metastatic colorectal cancer. Ann Surg 258:879–886

8. Guardino A, Cassidy M, Pienkowski T, Radulovic S, Legrand F, Nguyen A, Fernandez L, Coutts J, Moore N, Hwang O, Trieger

B, Brand L, Reiner L, Delcayre A, Godfrey W (2009) Results of two phase I clinical trials of MVA-BN-HER2 in HER-2 over-expressing metastatic breast cancer patients. Cancer Res 69(24 Suppl):5089

9. Mandl SJ, Rountree RB, Dalpozzo K, Do L, Lombardo JR, Schoonmaker PL, Dirmeier U, Steigerwald R, Giffon T, Laus R, Delcayre A (2012) Immunotherapy with MVA-BN(R)-HER2 induces HER-2-specific Th1 immunity and alters the intratu-moral balance of effector and regulatory T cells. Cancer Immu-nol Immunother 61:19–29

10. Penichet ML, Challita PM, Shin SU, Sampogna SL, Rosenb-latt JD, Morrison SL (1999) In vivo properties of three human HER2/neu-expressing murine cell lines in immunocompetent mice. Lab Anim Sci 49:179–188

11. Renard V, Sonderbye L, Ebbehoj K, Rasmussen PB, Gregorius K, Gottschalk T, Mouritsen S, Gautam A, Leach DR (2003) HER-2 DNA and protein vaccines containing potent Th cell epitopes induce distinct protective and therapeutic antitumor responses in HER-2 transgenic mice. J Immunol 171:1588–1595

12. Li Z, Ling L, Liu X, Laus R, Delcayre A (2010) A flow cytom-etry-based immuno-titration assay for rapid and accurate titer determination of Modified Vaccinia Ankara virus vectors. J Virol Methods 169:87–94

13. Betts MR, Brenchley JM, Price DA, De Rosa SC, Douek DC, Roederer M, Koup RA (2003) Sensitive and viable identification of antigen-specific CD8+ T cells by a flow cytometric assay for degranulation. J Immunol Methods 281:65–78

14. Tscharke DC, Woo WP, Sakala IG, Sidney J, Sette A, Moss DJ, Bennink JR, Karupiah G, Yewdell JW (2006) Poxvirus CD8+ T-cell determinants and cross-reactivity in BALB/c mice. J Virol 80:6318–6323

15. Okugawa T, Ikuta Y, Takahashi Y, Obata H, Tanida K, Watanabe M, Imai S, Furugen R, Nagata Y, Toyoda N, Shiku H (2000) A novel human HER2-derived peptide homologous to the mouse K(d)-restricted tumor rejection antigen can induce HLA-A24-re-stricted cytotoxic T lymphocytes in ovarian cancer patients and healthy individuals. Eur J Immunol 30:3338–3346

16. Pavlenko M, Leder C, Roos AK, Levitsky V, Pisa P (2005) Identification of an immunodominant H-2D(b)-restricted CTL epitope of human PSA. Prostate 64:50–59

17. Chou TC (2006) Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol Rev 58:621–681

18. Joshi NS, Cui W, Chandele A, Lee HK, Urso DR, Hagman J, Gapin L, Kaech SM (2007) Inflammation directs memory pre-cursor and short-lived effector CD8(+) T cell fates via the graded expression of T-bet transcription factor. Immunity 27:281–295

19. Olson JA, McDonald-Hyman C, Jameson SC, Hamilton SE (2013) Effector-like CD8(+) T cells in the memory population mediate potent protective immunity. Immunity 38:1250–1260

20. Carthon BC, Wolchok JD, Yuan J, Kamat A, Ng Tang DS, Sun J, Ku G, Troncoso P, Logothetis CJ, Allison JP, Sharma P (2010) Preoperative CTLA-4 blockade: tolerability and immune moni-toring in the setting of a presurgical clinical trial. Clin Cancer Res 16:2861–2871

21. Chen Y, Shen S, Gorentla BK, Gao J, Zhong XP (2012) Murine regulatory T cells contain hyperproliferative and death-prone subsets with differential ICOS expression. J Immunol 188:1698–1707

22. Dong C, Juedes AE, Temann UA, Shresta S, Allison JP, Ruddle NH, Flavell RA (2001) ICOS co-stimulatory receptor is essential for T-cell activation and function. Nature 409:97–101

23. Mandl SJ, Rountree RB, Dela Cruz TB, Foy SP, Cote JJ, Gordon EJ, Trent E, Delcayre A, Franzusoff A (2014) Elucidating immu-nologic mechanisms of PROSTVAC cancer immunotherapy. J Immunother Cancer 2:34

Page 13: Poxvirus-based active immunotherapy synergizes with CTLA-4 … · Cancer Immunol Immunother (2016) 65:537–549 DOI 10.1007/s00262-016-1816-7 ORIGINAL ARTICLE Poxvirus‑based active

549Cancer Immunol Immunother (2016) 65:537–549

1 3

24. Lauterbach H, Patzold J, Kassub R, Bathke B, Brinkmann K, Chaplin P, Suter M, Hochrein H (2013) Genetic adjuvantation of recombinant MVA with CD40L potentiates CD8 T cell mediated immunity. Front Immunol 4:251

25. Precopio ML, Betts MR, Parrino J, Price DA, Gostick E, Ambro-zak DR, Asher TE, Douek DC, Harari A, Pantaleo G, Bailer R, Graham BS, Roederer M, Koup RA (2007) Immunization with vaccinia virus induces polyfunctional and phenotypically distinc-tive CD8(+) T cell responses. J Exp Med 204:1405–1416

26. Seder RA, Darrah PA, Roederer M (2008) T-cell quality in mem-ory and protection: implications for vaccine design. Nat Rev Immunol 8:247–258

27. Fu T, He Q, Sharma P (2011) The ICOS/ICOSL pathway is required for optimal antitumor responses mediated by anti-CTLA-4 therapy. Cancer Res 71:5445–5454

28. Di Giacomo AM, Calabro L, Danielli R, Fonsatti E, Bertocci E, Pesce I, Fazio C, Cutaia O, Giannarelli D, Miracco C, Bia-gioli M, Altomonte M, Maio M (2013) Long-term survival and immunological parameters in metastatic melanoma patients who responded to ipilimumab 10 mg/kg within an expanded access programme. Cancer Immunol Immunother 62:1021–1028

29. Hutloff A, Dittrich AM, Beier KC, Eljaschewitsch B, Kraft R, Anagnostopoulos I, Kroczek RA (1999) ICOS is an induc-ible T-cell co-stimulator structurally and functionally related to CD28. Nature 397:263–266

30. Quezada SA, Peggs KS, Curran MA, Allison JP (2006) CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells. J Clin Invest 116:1935–1945

31. Liakou CI, Kamat A, Tang DN, Chen H, Sun J, Troncoso P, Logothetis C, Sharma P (2008) CTLA-4 blockade increases IFNgamma-producing CD4+ ICOShi cells to shift the ratio of effector to regulatory T cells in cancer patients. Proc Natl Acad Sci USA 105:14987–14992

32. Vonderheide RH, LoRusso PM, Khalil M, Gartner EM, Khaira D, Soulieres D, Dorazio P, Trosko JA, Ruter J, Mariani GL, Usari T, Domchek SM (2010) Tremelimumab in combina-tion with exemestane in patients with advanced breast cancer and treatment-associated modulation of inducible costimulator expression on patient T cells. Clin Cancer Res 16:3485–3494

33. Selby MJ, Engelhardt JJ, Quigley M, Henning KA, Chen T, Srin-ivasan M, Korman AJ (2013) Anti-CTLA-4 antibodies of IgG2a isotype enhance antitumor activity through reduction of intratu-moral regulatory T cells. Cancer Immunol Res 1:32–42

34. Kudo-Saito C, Schlom J, Hodge JW (2005) Induction of an antigen cascade by diversified subcutaneous/intratumoral vacci-nation is associated with antitumor responses. Clin Cancer Res 11:2416–2426

35. Gulley JL, Madan RA, Tsang KY, Jochems C, Marte JL, Farsaci B, Tucker JA, Hodge JW, Liewehr DJ, Steinberg SM, Heery CR, Schlom J (2014) Immune impact induced by PROSTVAC (PSA-TRICOM), a therapeutic vaccine for prostate cancer. Cancer Immunol Res 2:133–141

36. Arlen PM, Gulley JL, Parker C, Skarupa L, Pazdur M, Panicali D, Beetham P, Tsang KY, Grosenbach DW, Feldman J, Steinberg SM, Jones E, Chen C, Marte J, Schlom J, Dahut W (2006) A ran-domized phase II study of concurrent docetaxel plus vaccine ver-sus vaccine alone in metastatic androgen-independent prostate cancer. Clin Cancer Res 12:1260–1269

37. Kass E, Schlom J, Thompson J, Guadagni F, Graziano P, Greiner JW (1999) Induction of protective host immunity to carcinoem-bryonic antigen (CEA), a self-antigen in CEA transgenic mice, by immunizing with a recombinant vaccinia-CEA virus. Cancer Res 59:676–683

38. Aarts WM, Schlom J, Hodge JW (2002) Vector-based vaccine/cytokine combination therapy to enhance induction of immune responses to a self-antigen and antitumor activity. Cancer Res 62:5770–5777

39. Espenschied J, Lamont J, Longmate J, Pendas S, Wang Z, Dia-mond DJ, Ellenhorn JD (2003) CTLA-4 blockade enhances the therapeutic effect of an attenuated poxvirus vaccine targeting p53 in an established murine tumor model. J Immunol 170:3401–3407

40. Chakraborty M, Schlom J, Hodge JW (2007) The combined activation of positive costimulatory signals with modulation of a negative costimulatory signal for the enhancement of vac-cine-mediated T-cell responses. Cancer Immunol Immunother 56:1471–1484

41. Hodge JW, Chakraborty M, Kudo-Saito C, Garnett CT, Schlom J (2005) Multiple costimulatory modalities enhance CTL avidity. J Immunol 174:5994–6004

42. Gerritsen WR, Kwon ED, Fizazi K, Bossi A, Van den Eertwegh A, Logothetis C, Scher HI, Beer TM, McHenry MB, Gagnier P, Drake CG (2013) CA184-043: a randomized, multicenter, double-blind phase 3 trial comparing overall survival (OS) in patients (pts) with post-docetaxel castration-resistant prostate cancer (CRPC) and bone metastases treated with ipilimumab (ipi) vs placebo (pbo), each following single-dose radiotherapy (RT). Eur J Cancer 49(2 Suppl):S678